Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Neuron ; 103(1): 52-65.e6, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31101394

ABSTRACT

Mitochondria are essential in long axons to provide metabolic support and sustain neuron integrity. A healthy mitochondrial pool is maintained by biogenesis, transport, mitophagy, fission, and fusion, but how these events are regulated in axons is not well defined. Here, we show that the Drosophila glutathione S-transferase (GST) Gfzf prevents mitochondrial hyperfusion in axons. Gfzf loss altered redox balance between glutathione (GSH) and oxidized glutathione (GSSG) and initiated mitochondrial fusion through the coordinated action of Mfn and Opa1. Gfzf functioned epistatically with the thioredoxin peroxidase Jafrac1 and the thioredoxin reductase 1 TrxR-1 to regulate mitochondrial dynamics. Altering GSH:GSSG ratios in mouse primary neurons in vitro also induced hyperfusion. Mitochondrial changes caused deficits in trafficking, the metabolome, and neuronal physiology. Changes in GSH and oxidative state are associated with neurodegenerative diseases like Alzheimer's. Our demonstration that GSTs are key in vivo regulators of axonal mitochondrial length and number provides a potential mechanistic link.


Subject(s)
Axons/physiology , Carrier Proteins/physiology , Glutathione/metabolism , Mitochondria/physiology , Animals , Axons/ultrastructure , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Female , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Neurons/metabolism , Oxidation-Reduction , Peroxidases/genetics , Peroxidases/physiology , Pregnancy , Primary Cell Culture , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 1/physiology
2.
Am J Physiol Lung Cell Mol Physiol ; 315(4): L545-L552, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30024305

ABSTRACT

Thioredoxin reductase-1 (TXNRD1) inhibition effectively activates nuclear factor (erythroid-derived 2)-like 2 (Nrf2) responses and attenuates lung injury in acute respiratory distress syndrome (ARDS) and bronchopulmonary dysplasia (BPD) models. Upon TXNRD1 inhibition, heme oxygenase-1 (HO-1) is disproportionally increased compared with Nrf2 target NADPH quinone oxidoreductase-1 (Nqo1). HO-1 has been investigated as a potential therapeutic target in both ARDS and BPD. TXNRD1 is predominantly expressed in airway epithelial cells; however, the mechanism of HO-1 induction by TXNRD1 inhibitors is unknown. We tested the hypothesis that TXNRD1 inhibition induces HO-1 via Nrf2-dependent mechanisms. Wild-type (WT), Nrf2KO1.3, and Nrf2KO2.2 cells were morphologically indistinguishable, indicating that Nrf2 can be deleted from murine-transformed club cells (mtCCs) using CRISPR/Cas9 gene editing. Hemin, a Nrf2-independent HO-1-inducing agent, significantly increased HO-1 expression in WT, Nrf2KO1.3, and Nrf2KO2.2. Auranofin (AFN) (0.5 µM) inhibited TXNRD1 activity by 50% and increased Nqo1 and Hmox1 mRNA levels by 6- and 24-fold, respectively, in WT cells. Despite similar levels of TXNRD1 inhibition, Nqo1 mRNA levels were not different between control and AFN-treated Nrf2KO1.3 and Nrf2KO2.2. AFN slightly increased Hmox1 mRNA levels in Nrf2KO1.3 and Nrf2KO2.2 cells compared with controls. AFN failed to increase HO-1 protein in Nrf2KO1.3 and Nrf2KO2.2 compared with a 36-fold increase in WT mtCCs. Our data indicate that Nrf2 is the primary mechanism by which TXNRD1 inhibitors increase HO-1 in lung epithelia. Future studies will use ARDS and BPD models to define the role of HO-1 in attenuation of lung injury by TXNRD1 inhibitors.


Subject(s)
Auranofin/pharmacology , Epithelial Cells/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/metabolism , Lung/enzymology , Membrane Proteins/metabolism , NF-E2-Related Factor 2/physiology , Thioredoxin Reductase 1/physiology , Animals , Antirheumatic Agents/pharmacology , Cells, Cultured , Epithelial Cells/drug effects , Heme Oxygenase-1/genetics , Lung/drug effects , Membrane Proteins/genetics , Mice , Mice, Inbred C3H , Mice, Knockout , Thioredoxin Reductase 1/antagonists & inhibitors
3.
Nat Commun ; 9(1): 1851, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29749372

ABSTRACT

The thioredoxin-1 (Trx1) system is an important contributor to cellular redox balance and is a sensor of energy and glucose metabolism. Here we show critical c-Myc-dependent activation of the Trx1 system during thymocyte and peripheral T-cell proliferation, but repression during T-cell quiescence. Deletion of thioredoxin reductase-1 (Txnrd1) prevents expansion the CD4-CD8- thymocyte population, whereas Txnrd1 deletion in CD4+CD8+ thymocytes does not affect further maturation and peripheral homeostasis of αßT cells. However, Txnrd1 is critical for expansion of the activated T-cell population during viral and parasite infection. Metabolomics show that TrxR1 is essential for the last step of nucleotide biosynthesis by donating reducing equivalents to ribonucleotide reductase. Impaired availability of 2'-deoxyribonucleotides induces the DNA damage response and cell cycle arrest of Txnrd1-deficient T cells. These results uncover a pivotal function of the Trx1 system in metabolic reprogramming of thymic and peripheral T cells and provide a rationale for targeting Txnrd1 in T-cell leukemia.


Subject(s)
Carrier Proteins/metabolism , Cell Proliferation/physiology , Cellular Reprogramming/physiology , DNA/biosynthesis , T-Lymphocytes/physiology , Thioredoxin Reductase 1/physiology , Thioredoxins/metabolism , Thioredoxins/physiology , Animals , Bone Marrow Transplantation , Cell Line , Deoxyribonucleotides/biosynthesis , Disease Models, Animal , Down-Regulation , Female , Humans , Leishmania major/immunology , Leishmania major/pathogenicity , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/parasitology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Male , Metabolomics , Mice, Inbred C57BL , Mice, Transgenic , Transplantation Chimera
4.
Lab Invest ; 96(3): 307-16, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26692290

ABSTRACT

We previously showed that histone deacetylase inhibitor (HDACi) and 5-azacytidine (AZA) treatment selectively induced cell death of esophageal cancer cells. The mechanisms of cancer selectivity, however, remained unclear. Here we examined whether the cancer selectivity of HDACi/AZA treatment is mediated by the thioredoxin (Trx) system and reactive oxygen species (ROS) in esophageal cancer cells. For this, we first analyzed human tissue specimens of 37 esophageal cancer patients by immunohistochemistry for Trx, Trx-interacting protein (TXNIP) and Trx reductase (TXNRD). This revealed a loss or at least reduction of nuclear Trx in esophageal cancer cells, compared with normal epithelial cells (P<0.001). Although no differences were observed for TXNIP, TXNRD was more frequently expressed in cancer cells (P<0.001). In the two main histotypes of esophageal squamous cell carcinomas (ESCCs, n=19) and esophageal adenomcarcinomas (EAC, n=16), similar Trx, TXNIP and TXNRD expression patterns were observed. Also in vitro, nuclear Trx was only detectable in non-neoplastic Het-1A cells, but not in OE21/ESCC or OE33/EAC cell lines. Moreover, the two cancer cell lines showed an increased Trx activity, being significant for OE21 (P=0.0237). After treatment with HDACi and/or AZA, ROS were exclusively increased in both cancer cell lines (P=0.048-0.017), with parallel decrease of Trx activity. This was variably accompanied by increased TXNIP levels upon AZA, MS-275 or MS-275/AZA treatment for 6 or 24 h in OE21, but not in Het-1A or OE33 cells. In summary, this study evaluated Trx and its associated proteins TXNIP and TXNRD for the first time in esophageal cancers. The analyses revealed an altered subcellular localization of Trx and strong upregulation of TXNRD in esophageal cancer cells. Moreover, HDACi and AZA disrupted Trx function and induced accumulation of ROS with subsequent apoptosis in esophageal cancer cells exclusively. Trx function is hence an important cellular mediator conferring non-neoplastic cell resistance for HDACi and/or AZA.


Subject(s)
Azacitidine/therapeutic use , Esophageal Neoplasms/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Thioredoxins/physiology , Adult , Aged , Aged, 80 and over , Carrier Proteins/physiology , Cell Line, Tumor , Epigenesis, Genetic , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Humans , Male , Middle Aged , Reactive Oxygen Species/metabolism , Thioredoxin Reductase 1/physiology
5.
Cancer Res ; 74(22): 6717-30, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25261240

ABSTRACT

Colorectal tumorigenesis is accompanied by the generation of oxidative stress, but how this controls tumor development is poorly understood. Here, we studied how the H2O2-reducing enzyme glutathione peroxidase 2 (GPx2) regulates H2O2 stress and differentiation in patient-derived "colonosphere" cultures. GPx2 silencing caused accumulation of radical oxygen species, sensitization to H2O2-induced apoptosis, and strongly reduced clone- and metastasis-forming capacity. Neutralization of radical oxygen species restored clonogenic capacity. Surprisingly, GPx2-suppressed cells also lacked differentiation potential and formed slow-growing undifferentiated tumors. GPx2 overexpression stimulated multilineage differentiation, proliferation, and tumor growth without reducing the tumor-initiating capacity. Finally, GPx2 expression was inversely correlated with H2O2-stress signatures in human colon tumor cohorts, but positively correlated with differentiation and proliferation. Moreover, high GPx2 expression was associated with early tumor recurrence, particularly in the recently identified aggressive subtype of human colon cancer. We conclude that H2O2 neutralization by GPx2 is essential for maintaining clonogenic and metastatic capacity, but also for the generation of differentiated proliferating tumor mass. The results reveal an unexpected redox-controlled link between tumor mass formation and metastatic capacity.


Subject(s)
Colorectal Neoplasms/pathology , Glutathione Peroxidase/physiology , Hydrogen Peroxide/metabolism , Animals , Cell Differentiation , Colorectal Neoplasms/metabolism , Female , Humans , Mice , Mice, SCID , Neoplasm Metastasis , Reactive Oxygen Species/metabolism , Stress, Physiological , Thioredoxin Reductase 1/physiology
6.
Carcinogenesis ; 33(9): 1806-13, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22791808

ABSTRACT

Thioredoxin reductase 1 (TR1) controls the redox state of protein thiols in mammalian cells and has been shown to have roles in both preventing and promoting cancer. To define the role of this selenoenzyme in hepatocellular carcinoma development, we examined tumor incidence in the liver of mice with tissue-specific knockout of mouse TR1 subjected to the liver carcinogen, diethylnitrosamine (DEN). TR1-deficient livers manifested ~90% tumor incidence compared with ~16% in control livers. The TR1-dependent effect was observed independent of sex, and, in control mice, tumorigenesis did not affect the expression of TR1. On the other hand, we observed upregulation of another selenoenzyme, glutathione peroxidase 2 (GPx2), and components of the glutathione (GSH) system, including those that generate reduced GSH. Overall, this study shows that TR1 protects against chemically induced hepatocarcinogenesis via the control of the cellular redox state, whereas its role in promoting this type of cancer is minimal.


Subject(s)
Liver Neoplasms/prevention & control , Thioredoxin Reductase 1/physiology , Animals , Body Weight , Female , Glutathione/metabolism , Glutathione Peroxidase/analysis , Homeostasis , Liver Neoplasms/chemically induced , Male , Mice , Mice, Inbred C57BL , Organ Size , Oxidation-Reduction , Glutathione Peroxidase GPX1
7.
Osteoarthritis Cartilage ; 20(5): 440-445, 2012 May.
Article in English | MEDLINE | ID: mdl-22353746

ABSTRACT

OBJECTIVE: Selenium neutralizes interleukin-1ß (IL-1ß) induced inflammatory responses in chondrocytes. We investigated potential mechanisms for this through in vitro knock down of three major selenoproteins, Iodothyronine Deiodinase-2 (DIO2), Glutathione Peroxidase-1 (GPX1), and Thioredoxin Reductase-1 (TR1) in primary human chondrocytes. METHODS: Primary human chondrocytes were transfected with scrambled small interfering ribonucleic acid (siRNA) or siRNA specific for DIO2, GPX1 and TR1. After 48 h, transfected cells were cultured in serum free media for 48 h, with or without 10 pg/ml IL-1ß for the final 24h. The efficiency of siRNAs was confirmed by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR) and Western blot analysis. The gene expression, by qRT-PCR, of cyclooxygenase-2 (COX2), IL-1ß, and Liver X receptor (LXR) alpha and beta was evaluated to determine the impact of selenoprotein knockdown on inflammatory responses in chondrocytes. RESULTS: The messenger RNA (mRNA) expression of DIO2, GPX1, and TR1 was significantly decreased by the specific siRNAs (reduced 56%, P=0.0004; 96%, P<0.0001; and 66%, P<0.0001, respectively). Suppression of DIO2, but not GPX1 or TR1, significantly increased (~2-fold) both basal (P=0.0005) and IL-1ß induced (P<0.0001) COX2 gene expression. Similarly, suppression of DIO2 significantly increased (∼9-fold) IL-1ß induced IL-1ß gene expression (P=0.0056) and resulted in a 32% (P=0.0044) decrease in LXRα gene expression but no effect on LXRß. CONCLUSIONS: Suppression of the selenoprotein DIO2 resulted in strong pro-inflammatory effects with increased expression of inflammatory mediators, IL-1ß and COX2, and decreased expression of LXRα suggesting that this may be the upstream target through which the anti-inflammatory effects of DIO2 are mediated.


Subject(s)
Chondrocytes/metabolism , Inflammation Mediators/metabolism , Iodide Peroxidase/physiology , Cells, Cultured , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Glutathione Peroxidase/genetics , Glutathione Peroxidase/physiology , Humans , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Iodide Peroxidase/genetics , Liver X Receptors , Orphan Nuclear Receptors/biosynthesis , Orphan Nuclear Receptors/genetics , RNA, Small Interfering/genetics , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 1/physiology , Transfection , Glutathione Peroxidase GPX1 , Iodothyronine Deiodinase Type II
8.
Toxicol Appl Pharmacol ; 241(3): 348-55, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19782697

ABSTRACT

The relationship between selenium and cancer is complex because individuals with low serum selenium levels benefit from selenium supplementation, but those with high serum selenium levels are at increased risk for other diseases. This suggests that the use of selenocompounds might be limited to particular circumstances, such as adjuvant therapy. A contributor to this dichotomy may be the activity of certain selenium containing enzymes like the cytosolic thioredoxin reductase (TR1). We evaluated the cellular response to select selenocompounds that have anticancer activity when TR1 was attenuated by siRNA in RKO colon cancer cells. Methylseleninic acid (MSA), which is a substrate for TR1, enhanced cytotoxicity to colon cancer cells when TR1 was attenuated. MSA induced stress in the endoplasmic reticulum, as measured by GRP78 protein levels. However, this pathway did not appear to account for the change in cytotoxicity when TR1 was attenuated. Instead, knockdown of the cytosolic TR plus incubation with MSA increased autophagy, as measured by LC3B cleavage, and apoptosis, as measured by Annexin V and mitochondrial dysfunction. Therefore, the use of selenocompounds with anticancer activity, like MSA, might be utilized most effectively with agents that targets TR1 in chemotherapeutic applications.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Organoselenium Compounds/toxicity , Thioredoxin Reductase 1/antagonists & inhibitors , Thioredoxin Reductase 1/physiology , Adenosine Triphosphate/metabolism , Annexin A5 , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/pathology , Endoplasmic Reticulum Chaperone BiP , Enzyme Inhibitors , Flow Cytometry , Humans , Immunohistochemistry , Membrane Potentials/drug effects , Mitochondria/drug effects , RNA, Small Interfering , Sulfhydryl Compounds/metabolism
9.
PLoS One ; 4(7): e6158, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19584930

ABSTRACT

BACKGROUND: Metabolically active cells require robust mechanisms to combat oxidative stress. The cytoplasmic thioredoxin reductase/thioredoxin (Txnrd1/Txn1) system maintains reduced protein dithiols and provides electrons to some cellular reductases, including peroxiredoxins. PRINCIPAL FINDINGS: Here we generated mice in which the txnrd1 gene, encoding Txnrd1, was specifically disrupted in all parenchymal hepatocytes. Txnrd1-deficient livers exhibited a transcriptome response in which 56 mRNAs were induced and 12 were repressed. Based on the global hybridization profile, this represented only 0.3% of the liver transcriptome. Since most liver mRNAs were unaffected, compensatory responses were evidently effective. Nuclear pre-mRNA levels indicated the response was transcriptional. Twenty-one of the induced genes contained known antioxidant response elements (AREs), which are binding sites for the oxidative and chemical stress-induced transcription factor Nrf2. Txnrd1-deficient livers showed increased accumulation of nuclear Nrf2 protein and chromatin immunoprecipitation on the endogenous nqo1 and aox1 promoters in fibroblasts indicated that Txnrd1 ablation triggered in vivo assembly of Nrf2 on each. CONCLUSIONS: Chronic deletion of Txnrd1 results in induction of the Nrf2 pathway, which contributes to an effective compensatory response.


Subject(s)
Hepatocytes/metabolism , NF-E2-Related Factor 2/metabolism , Thioredoxin Reductase 1/physiology , Animals , Cells, Cultured , Chromatin Immunoprecipitation , Gene Expression Profiling , Immunohistochemistry , Mice , Oxidation-Reduction , RNA, Messenger/genetics , Thioredoxin Reductase 1/genetics
10.
Biochim Biophys Acta ; 1790(11): 1541-5, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19272412

ABSTRACT

Of the many health benefits attributed to selenium, the one that has received the most attention is its role in cancer prevention. Selenium-containing proteins (selenoproteins) have been shown in recent years to have roles in cancer prevention. However, selenoproteins have diverse functions and their view as antioxidants is oversimplified. Some selenoproteins appear to have a split personality in having roles both in preventing and promoting cancer. The contrasting roles of one selenoprotein, thioredoxin reductase 1, in cancer are discussed in detail, but as also noted, at least one other selenoprotein may also have such a dual function. In addition, we discuss examples of inhibition of cancer development by selenoprotein deficiency in mouse models. These studies highlight the complex nature of selenium in relation to cancer.


Subject(s)
Neoplasms/etiology , Neoplasms/prevention & control , Selenoproteins/physiology , Animals , Cell Transformation, Neoplastic/genetics , Humans , Mice , Selenoproteins/deficiency , Selenoproteins/genetics , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 1/physiology
11.
Thromb Res ; 123(5): 727-30, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19128823

ABSTRACT

INTRODUCTION: Tetramethyl pyrazine has been considered an effective agent in treating neurons ischemia/reperfusion injury, but the mechanism of its therapeutic effect remains unclear. This study was to explore the therapeutic time window and mechanism of tetramethyl pyrazine on temporary focal cerebral ischemia/reperfusion injury. MATERIALS AND METHODS: Middle cerebral artery occlusion was conducted in male Sprague-Dawley rats and 20 mg/kg of tetramethyl pyrazine was intraperitoneally injected at different time points. At 72 h after reperfusion, all animals' neurologic deficit scores were evaluated. Cerebrums were removed and cerebral infarction volume was measured. The expression of thioredoxin and thioredoxin reductase mRNA was determined at 6 and 24 h after reperfusion. RESULTS: Cerebral infarction volume and neurological deficit scores were significantly decreased in the group with tetramethyl pyrazine treatment. The expression of thioredoxin-1/thioredoxin-2 and thioredoxin reductase-1/thioredoxin reductase-2 was significantly decreased in rats with ischemia/reperfusion injury, while it was increased by tetramethyl pyrazine administration. CONCLUSIONS: Treatment with tetramethyl pyrazine, within 4 h after reperfusion, protects the brain from ischemic reperfusion injury in rats. The neuroprotective mechanism of tetramethyl pyrazine treatment is, in part, mediated through the upregulation of thioredoxin transcription.


Subject(s)
Brain Ischemia/drug therapy , Neuroprotective Agents/therapeutic use , Pyrazines/therapeutic use , Reperfusion Injury/prevention & control , Animals , Apoptosis/drug effects , Male , Pyrazines/pharmacology , Rats , Rats, Sprague-Dawley , Thioredoxin Reductase 1/physiology , Thioredoxins/physiology
12.
Biochem Biophys Res Commun ; 371(1): 63-8, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18406344

ABSTRACT

Intracellular levels of iron are tightly regulated. Saccharomyces cerevisiae uses well-defined pathways to extract iron molecules from the environment. Once inside the cell, the iron molecules must be transferred to target sites via an intracellular iron transporter. Although analogous carriers have been described for other metals, such as copper, an iron transporter has yet to be identified. We used two-dimensional gel electrophoresis and mass spectrometry techniques to attempt to identify the iron transporter from cytosolic fraction of S. cerevisiae. In this study, we identified the iron-binding activity of thioredoxin reductase, and our data suggest a potential role for this enzyme in intracellular iron transport.


Subject(s)
Iron-Binding Proteins/physiology , Iron/metabolism , Saccharomyces cerevisiae/enzymology , Thioredoxin Reductase 1/physiology , Chromatography, Affinity/methods , Circular Dichroism , Culture Media , Iron-Binding Proteins/genetics , Iron-Binding Proteins/isolation & purification , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Spectrophotometry, Ultraviolet , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 1/isolation & purification
13.
J Biol Chem ; 283(5): 2814-21, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18042542

ABSTRACT

The human thioredoxin system has a wide range of functions in cells including regulation of cell proliferation and differentiation, immune system modulation, antioxidant defense, redox control of transcription factor activity, and promotion of cancer development. A key component of this enzymatic system is the selenoprotein thioredoxin reductase 1 (TrxR1), encoded by the TXNRD1 gene. Transcription of TXNRD1 involves alternative splicing, leading to a number of transcripts also encoding isoforms of TrxR1 that differ from each other at their N-terminal domains. Here we have studied the TXNRD1_v3 isoform containing an atypical N-terminal glutaredoxin (Grx) domain. Expression of the transcript of this isoform was found predominantly in testis but was also detected in ovary, spleen, heart, liver, kidney, and pancreas. By immunohistochemical analysis in human testis with antibodies specific for the Grx domain of TXNRD1_v3, the protein was found to be predominantly expressed in the Leydig cells. Expression of the TXNRD1_v3 transcript was also found in several cancer cell lines (HCC1937, H23, A549, U1810, or H157), and in HeLa cells, it was induced by estradiol or testosterone treatments. Surprisingly, green fluorescent protein fusions with the complete TXNRD1_v3 protein or with only its Grx domain localized to distinct cellular sites in proximity to actin, and furthermore, had a potent capacity to rapidly induce cell membrane protrusions. Analyses of these structures suggested that the Grx domain of TXNRD1_v3 localizes first in the emerging protrusion and is then followed into the protrusions by actin and subsequently by tubulin. The results presented thus reveal that TXNRD1_v3 has a unique and distinct expression pattern in human cells and suggest that the protein can guide actin polymerization in relation to cell membrane restructuring.


Subject(s)
Cell Membrane/enzymology , Cell Membrane/ultrastructure , Thioredoxin Reductase 1/chemistry , Thioredoxin Reductase 1/physiology , Actins/metabolism , Alternative Splicing , Base Sequence , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , DNA Primers/genetics , Glutaredoxins/chemistry , HeLa Cells , Humans , Male , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Testis/enzymology , Testis/ultrastructure , Thioredoxin Reductase 1/genetics , Transfection , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...